51
|
Pichaud F. PAR-Complex and Crumbs Function During Photoreceptor Morphogenesis and Retinal Degeneration. Front Cell Neurosci 2018; 12:90. [PMID: 29651238 PMCID: PMC5884931 DOI: 10.3389/fncel.2018.00090] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/15/2018] [Indexed: 12/30/2022] Open
Abstract
The fly photoreceptor has long been used as a model to study sensory neuron morphogenesis and retinal degeneration. In particular, elucidating how these cells are built continues to help further our understanding of the mechanisms of polarized cell morphogenesis, intracellular trafficking and the causes of human retinal pathologies. The conserved PAR complex, which in flies consists of Cdc42-PAR6-aPKC-Bazooka, and the transmembrane protein Crumbs (Crb) are key players during photoreceptor morphogenesis. While the PAR complex regulates polarity in many cell types, Crb function in polarity is relatively specific to epithelial cells. Together Cdc42-PAR6-aPKC-Bazooka and Crb orchestrate the differentiation of the photoreceptor apical membrane (AM) and zonula adherens (ZA), thus allowing these cells to assemble into a neuro-epithelial lattice. In addition to its function in epithelial polarity, Crb has also been shown to protect fly photoreceptors from light-induced degeneration, a process linked to Rhodopsin expression and trafficking. Remarkably, mutations in the human Crumbs1 (CRB1) gene lead to retinal degeneration, making the fly photoreceptor a powerful disease model system.
Collapse
Affiliation(s)
- Franck Pichaud
- Medical Research Council, Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| |
Collapse
|
52
|
Ashley J, Cordy B, Lucia D, Fradkin LG, Budnik V, Thomson T. Retrovirus-like Gag Protein Arc1 Binds RNA and Traffics across Synaptic Boutons. Cell 2018; 172:262-274.e11. [PMID: 29328915 PMCID: PMC5793882 DOI: 10.1016/j.cell.2017.12.022] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 08/15/2017] [Accepted: 12/18/2017] [Indexed: 12/31/2022]
Abstract
Arc/Arg3.1 is required for synaptic plasticity and cognition, and mutations in this gene are linked to autism and schizophrenia. Arc bears a domain resembling retroviral/retrotransposon Gag-like proteins, which multimerize into a capsid that packages viral RNA. The significance of such a domain in a plasticity molecule is uncertain. Here, we report that the Drosophila Arc1 protein forms capsid-like structures that bind darc1 mRNA in neurons and is loaded into extracellular vesicles that are transferred from motorneurons to muscles. This loading and transfer depends on the darc1-mRNA 3' untranslated region, which contains retrotransposon-like sequences. Disrupting transfer blocks synaptic plasticity, suggesting that transfer of dArc1 complexed with its mRNA is required for this function. Notably, cultured cells also release extracellular vesicles containing the Gag region of the Copia retrotransposon complexed with its own mRNA. Taken together, our results point to a trans-synaptic mRNA transport mechanism involving retrovirus-like capsids and extracellular vesicles.
Collapse
Affiliation(s)
- James Ashley
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Benjamin Cordy
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Diandra Lucia
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lee G Fradkin
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Vivian Budnik
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Travis Thomson
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
53
|
Riedel F, Galindo A, Muschalik N, Munro S. The two TRAPP complexes of metazoans have distinct roles and act on different Rab GTPases. J Cell Biol 2017; 217:601-617. [PMID: 29273580 PMCID: PMC5800803 DOI: 10.1083/jcb.201705068] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/19/2017] [Accepted: 11/27/2017] [Indexed: 12/03/2022] Open
Abstract
In yeast, the TRAPP complexes activate Rab1 with TRAPPII also activating Rab11, but less is known about the two TRAPPs in metazoans. Riedel et al. show that in Drosophila melanogaster, TRAPPIII is an essential Rab1 activator, and TRAPPII activates Rab1 and Rab11 and becomes essential when an unrelated Rab11 activator is deleted. Originally identified in yeast, transport protein particle (TRAPP) complexes are Rab GTPase exchange factors that share a core set of subunits. TRAPPs were initially found to act on Ypt1, the yeast orthologue of Rab1, but recent studies have found that yeast TRAPPII can also activate the Rab11 orthologues Ypt31/32. Mammals have two TRAPP complexes, but their role is less clear, and they contain subunits that are not found in the yeast complexes but are essential for cell growth. To investigate TRAPP function in metazoans, we show that Drosophila melanogaster have two TRAPP complexes similar to those in mammals and that both activate Rab1, whereas one, TRAPPII, also activates Rab11. TRAPPII is not essential but becomes so in the absence of the gene parcas that encodes the Drosophila orthologue of the SH3BP5 family of Rab11 guanine nucleotide exchange factors (GEFs). Thus, in metazoans, Rab1 activation requires TRAPP subunits not found in yeast, and Rab11 activation is shared by TRAPPII and an unrelated GEF that is metazoan specific.
Collapse
Affiliation(s)
- Falko Riedel
- Medical Research Council Laboratory of Molecular Biology, Cambridge, England, UK
| | - Antonio Galindo
- Medical Research Council Laboratory of Molecular Biology, Cambridge, England, UK
| | - Nadine Muschalik
- Medical Research Council Laboratory of Molecular Biology, Cambridge, England, UK
| | - Sean Munro
- Medical Research Council Laboratory of Molecular Biology, Cambridge, England, UK
| |
Collapse
|
54
|
Abstract
Polarized exocytosis is generally considered as the multistep vesicular trafficking process in which membrane-bounded carriers are transported from the Golgi or endosomal compartments to specific sites of the plasma membrane. Polarized exocytosis in cells is achieved through the coordinated actions of membrane trafficking machinery and cytoskeleton orchestrated by signaling molecules such as the Rho family of small GTPases. Elucidating the molecular mechanisms of polarized exocytosis is essential to our understanding of a wide range of pathophysiological processes from neuronal development to tumor invasion.
Collapse
Affiliation(s)
- Jingwen Zeng
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6018
| | - Shanshan Feng
- Key Laboratory for Regenerative Medicine of Ministry of Education and Department of Developmental & Regenerative Biology, Jinan University, Guangzhou 510632, P.R. China
| | - Bin Wu
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6018
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6018
| |
Collapse
|
55
|
Del Signore SJ, Biber SA, Lehmann KS, Heimler SR, Rosenfeld BH, Eskin TL, Sweeney ST, Rodal AA. dOCRL maintains immune cell quiescence by regulating endosomal traffic. PLoS Genet 2017; 13:e1007052. [PMID: 29028801 PMCID: PMC5656325 DOI: 10.1371/journal.pgen.1007052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 10/25/2017] [Accepted: 10/04/2017] [Indexed: 01/07/2023] Open
Abstract
Lowe Syndrome is a developmental disorder characterized by eye, kidney, and neurological pathologies, and is caused by mutations in the phosphatidylinositol-5-phosphatase OCRL. OCRL plays diverse roles in endocytic and endolysosomal trafficking, cytokinesis, and ciliogenesis, but it is unclear which of these cellular functions underlie specific patient symptoms. Here, we show that mutation of Drosophila OCRL causes cell-autonomous activation of hemocytes, which are macrophage-like cells of the innate immune system. Among many cell biological defects that we identified in docrl mutant hemocytes, we pinpointed the cause of innate immune cell activation to reduced Rab11-dependent recycling traffic and concomitantly increased Rab7-dependent late endosome traffic. Loss of docrl amplifies multiple immune-relevant signals, including Toll, Jun kinase, and STAT, and leads to Rab11-sensitive mis-sorting and excessive secretion of the Toll ligand Spåtzle. Thus, docrl regulation of endosomal traffic maintains hemocytes in a poised, but quiescent state, suggesting mechanisms by which endosomal misregulation of signaling may contribute to symptoms of Lowe syndrome. Lowe syndrome is a developmental disorder characterized by severe kidney, eye, and neurological symptoms, and is caused by mutations in the gene OCRL. OCRL has been shown to control many steps of packaging and transport of materials within cells, though it remains unclear which of these disrupted transport steps cause each of the many symptoms in Lowe syndrome patients. We found that in fruit flies, loss of OCRL caused transport defects at specific internal compartments in innate immune cells, resulting in amplification of multiple critical inflammatory signals. Similar inflammatory signals have been implicated in forms of epilepsy, which is a primary symptom in Lowe syndrome patients. Thus, our work uncovers a new function for OCRL in animals, and opens an exciting new avenue of investigation into how loss of OCRL causes the symptoms of Lowe syndrome.
Collapse
Affiliation(s)
- Steven J. Del Signore
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Sarah A. Biber
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Katherine S. Lehmann
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Stephanie R. Heimler
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Benjamin H. Rosenfeld
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Tania L. Eskin
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Sean T. Sweeney
- Department of Biology, University of York, York, United Kingdom
| | - Avital A. Rodal
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
56
|
Wang J, Fresquez T, Kandachar V, Deretic D. The Arf GEF GBF1 and Arf4 synergize with the sensory receptor cargo, rhodopsin, to regulate ciliary membrane trafficking. J Cell Sci 2017; 130:3975-3987. [PMID: 29025970 DOI: 10.1242/jcs.205492] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 10/10/2017] [Indexed: 01/05/2023] Open
Abstract
The small GTPase Arf4 and the Arf GTPase-activating protein (GAP) ASAP1 cooperatively sequester sensory receptor cargo into transport carriers targeted to primary cilia, but the input that drives Arf4 activation in this process remains unknown. Here, we show, by using frog retinas and recombinant human proteins, that during the carrier biogenesis from the photoreceptor Golgi/trans-Golgi network (TGN) a functional complex is formed between Arf4, the Arf guanine nucleotide exchange factor (GEF) GBF1 and the light-sensing receptor, rhodopsin. Rhodopsin and Arf4 bind the regulatory N-terminal dimerization and cyclophillin-binding (DCB)-homology upstream of Sec7 (HUS) domain of GBF1. The complex is sensitive to Golgicide A (GCA), a selective inhibitor of GBF1 that accordingly blocks rhodopsin delivery to the cilia, without disrupting the photoreceptor Golgi. The emergence of newly synthesized rhodopsin in the endomembrane system is essential for GBF1-Arf4 complex formation in vivo Notably, GBF1 interacts with the Arf GAP ASAP1 in a GCA-resistant manner. Our findings indicate that converging signals on GBF1 from the influx of cargo into the Golgi/TGN and the feedback from Arf4, combined with input from ASAP1, control Arf4 activation during sensory membrane trafficking to primary cilia.
Collapse
Affiliation(s)
- Jing Wang
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - Theresa Fresquez
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - Vasundhara Kandachar
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - Dusanka Deretic
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, New Mexico 87131, USA .,Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico 87131, USA
| |
Collapse
|
57
|
Augustin H, McGourty K, Allen MJ, Madem SK, Adcott J, Kerr F, Wong CT, Vincent A, Godenschwege T, Boucrot E, Partridge L. Reduced insulin signaling maintains electrical transmission in a neural circuit in aging flies. PLoS Biol 2017; 15:e2001655. [PMID: 28902870 PMCID: PMC5597081 DOI: 10.1371/journal.pbio.2001655] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 08/04/2017] [Indexed: 12/11/2022] Open
Abstract
Lowered insulin/insulin-like growth factor (IGF) signaling (IIS) can extend healthy lifespan in worms, flies, and mice, but it can also have adverse effects (the “insulin paradox”). Chronic, moderately lowered IIS rescues age-related decline in neurotransmission through the Drosophila giant fiber system (GFS), a simple escape response neuronal circuit, by increasing targeting of the gap junctional protein innexin shaking-B to gap junctions (GJs). Endosomal recycling of GJs was also stimulated in cultured human cells when IIS was reduced. Furthermore, increasing the activity of the recycling small guanosine triphosphatases (GTPases) Rab4 or Rab11 was sufficient to maintain GJs upon elevated IIS in cultured human cells and in flies, and to rescue age-related loss of GJs and of GFS function. Lowered IIS thus elevates endosomal recycling of GJs in neurons and other cell types, pointing to a cellular mechanism for therapeutic intervention into aging-related neuronal disorders. Insulin and insulin-like growth factors play an important role in the nervous system development and function. Reduced insulin signaling, however, can improve symptoms of neurodegenerative diseases in different model organisms and protect against age-associated decline in neuronal function extending lifespan. Here, we analyze the effects of genetically attenuated insulin signaling on the escape response pathway in the fruit fly Drosophila melanogaster. This simple neuronal circuit is dominated by electrical synapses composed of the gap junctional shaking-B protein, which allows for the transfer of electrical impulses between cells. Transmission through the circuit is known to slow down with age. We show that this functional decline is prevented by systemic or circuit-specific suppression of insulin signaling due to the preservation of the number of gap junctional proteins in aging animals. Our experiments in a human cell culture system reveal increased membrane targeting of gap junctional proteins via small proteins Rab4 and Rab11 under reduced insulin conditions. We also find that increasing the level of these recycling-mediating proteins in flies preserves the escape response circuit output in old flies and suggests ways of improving the function of neuronal circuits dominated by electrical synapses during aging.
Collapse
Affiliation(s)
- Hrvoje Augustin
- Max Planck Institute for Biology of Aging, Köln, Germany
- Institute of Healthy Aging, and Genetics, Evolution, and Environment, University College London, London, United Kingdom
| | - Kieran McGourty
- Department of Structural and Molecular Biology, London, United Kingdom
| | - Marcus J. Allen
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | - Sirisha Kudumala Madem
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Jennifer Adcott
- Max Planck Institute for Biology of Aging, Köln, Germany
- Institute of Healthy Aging, and Genetics, Evolution, and Environment, University College London, London, United Kingdom
| | - Fiona Kerr
- Max Planck Institute for Biology of Aging, Köln, Germany
- Institute of Healthy Aging, and Genetics, Evolution, and Environment, University College London, London, United Kingdom
| | - Chi Tung Wong
- Max Planck Institute for Biology of Aging, Köln, Germany
| | - Alec Vincent
- Max Planck Institute for Biology of Aging, Köln, Germany
| | - Tanja Godenschwege
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Emmanuel Boucrot
- Department of Structural and Molecular Biology, London, United Kingdom
| | - Linda Partridge
- Max Planck Institute for Biology of Aging, Köln, Germany
- Institute of Healthy Aging, and Genetics, Evolution, and Environment, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
58
|
Giglio A, Perrotta ID, Brandmayr P. Exosomes: Ultrastructural evidence in epithelial cells of Malpighian tubules. Micron 2017; 100:34-37. [DOI: 10.1016/j.micron.2017.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 12/13/2022]
|
59
|
Chinchore Y, Begaj T, Wu D, Drokhlyansky E, Cepko CL. Glycolytic reliance promotes anabolism in photoreceptors. eLife 2017; 6. [PMID: 28598329 PMCID: PMC5499945 DOI: 10.7554/elife.25946] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 06/01/2017] [Indexed: 12/18/2022] Open
Abstract
Vertebrate photoreceptors are among the most metabolically active cells, exhibiting a high rate of ATP consumption. This is coupled with a high anabolic demand, necessitated by the diurnal turnover of a specialized membrane-rich organelle, the outer segment, which is the primary site of phototransduction. How photoreceptors balance their catabolic and anabolic demands is poorly understood. Here, we show that rod photoreceptors in mice rely on glycolysis for their outer segment biogenesis. Genetic perturbations targeting allostery or key regulatory nodes in the glycolytic pathway impacted the size of the outer segments. Fibroblast growth factor signaling was found to regulate glycolysis, with antagonism of this pathway resulting in anabolic deficits. These data demonstrate the cell autonomous role of the glycolytic pathway in outer segment maintenance and provide evidence that aerobic glycolysis is part of a metabolic program that supports the biosynthetic needs of a normal neuronal cell type. DOI:http://dx.doi.org/10.7554/eLife.25946.001 Living cells need building materials and energy to grow and carry out their activities. Most cells in the body use sugars like glucose for these purposes. In a process known as glycolysis, cells break down glucose into molecules that are eventually converted to carbon dioxide and water to form the chemical ATP – the cellular currency for energy. Developing cells that have not yet fully specialized, and rapidly dividing cells, like cancer cells, consume large amounts of glucose via aerobic glycolysis (also known as the Warburg effect) as they require high levels of energy and building materials. As cells become more specialized and divide less often, they have a reduced need for building blocks, and adjust their consumption and breakdown of glucose accordingly. One exception is the photoreceptor cells, found in the light-sensitive part of our eyes. Although these specialized cells do not divide, they still need a lot of energy and building blocks to constantly renew their light-sensing and processing structures, and to capture and convert the information from the environment into signals. Previous research has shown that the eye also uses the Warburg effect. However, until now, it was not known whether the photoreceptors or other cells in the eye carry out this form of glycolysis. Using genetic tools, Chinchore et al. analysed how the photoreceptor cells in mice used glucose. The experiments demonstrated that the photoreceptors do indeed carry out the Warburg effect. Chinchore et al. further discovered that the Warburg effect is regulated by the same key enzymes and signalling molecules that cancer cells use. This indicates that specialized cells like photoreceptors might choose to retain certain metabolic features of their precursor cells, if they need to. These findings provide new insight into how photoreceptors use glucose. The next step will be to understand how aerobic glycolysis is regulated in healthy eyes as well as in eyes that are affected by degenerating diseases, which may ultimately lead to new ways of treating blindness. DOI:http://dx.doi.org/10.7554/eLife.25946.002
Collapse
Affiliation(s)
- Yashodhan Chinchore
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, United States
| | - Tedi Begaj
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, United States
| | - David Wu
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, United States
| | - Eugene Drokhlyansky
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, United States
| | - Constance L Cepko
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| |
Collapse
|
60
|
Kamalesh K, Trivedi D, Toscano S, Sharma S, Kolay S, Raghu P. Phosphatidylinositol 5-phosphate 4-kinase regulates early endosomal dynamics during clathrin-mediated endocytosis. J Cell Sci 2017; 130:2119-2133. [PMID: 28507272 PMCID: PMC5536888 DOI: 10.1242/jcs.202259] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/09/2017] [Indexed: 12/19/2022] Open
Abstract
Endocytic turnover is essential for the regulation of the protein composition and function of the plasma membrane, and thus affects the plasma membrane levels of many receptors. In Drosophila melanogaster photoreceptors, photon absorption by the G-protein-coupled receptor (GPCR) rhodopsin 1 (Rh1; also known as NinaE) triggers its endocytosis through clathrin-mediated endocytosis (CME). We find that CME of Rh1 is regulated by phosphatidylinositol 5 phosphate 4-kinase (PIP4K). Flies lacking PIP4K show mislocalization of Rh1 on expanded endomembranes within the cell body. This mislocalization of Rh1 was dependent on the formation of an expanded Rab5-positive compartment. The Rh1-trafficking defect in PIP4K-depleted cells could be suppressed by downregulating Rab5 function or by selectively reconstituting PIP4K in the PI3P-enriched early endosomal compartment of photoreceptors. We also found that loss of PIP4K was associated with increased CME and an enlarged Rab5-positive compartment in cultured Drosophila cells. Collectively, our findings define PIP4K as a novel regulator of early endosomal homeostasis during CME.
Collapse
Affiliation(s)
- Kumari Kamalesh
- National Centre for Biological Sciences-TIFR, GKVK Campus, Bellary Road, Bangalore 560065, India.,Department of Biological Sciences, Tata Institute of Fundamental Research, Dr. Homi Bhabha Road, Colaba, Mumbai 400005, India
| | - Deepti Trivedi
- National Centre for Biological Sciences-TIFR, GKVK Campus, Bellary Road, Bangalore 560065, India
| | - Sarah Toscano
- Inositide Laboratory, Babraham Institute, Cambridge CB22 3AT, UK
| | - Sanjeev Sharma
- National Centre for Biological Sciences-TIFR, GKVK Campus, Bellary Road, Bangalore 560065, India
| | - Sourav Kolay
- National Centre for Biological Sciences-TIFR, GKVK Campus, Bellary Road, Bangalore 560065, India.,Manipal University, Madhav Nagar, Manipal, Karnataka 576104, India
| | - Padinjat Raghu
- National Centre for Biological Sciences-TIFR, GKVK Campus, Bellary Road, Bangalore 560065, India
| |
Collapse
|
61
|
Astrocyte Ca 2+ Influx Negatively Regulates Neuronal Activity. eNeuro 2017; 4:eN-NWR-0340-16. [PMID: 28303263 PMCID: PMC5348542 DOI: 10.1523/eneuro.0340-16.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/13/2017] [Accepted: 02/28/2017] [Indexed: 02/07/2023] Open
Abstract
Maintenance of neural circuit activity requires appropriate regulation of excitatory and inhibitory synaptic transmission. Recently, glia have emerged as key partners in the modulation of neuronal excitability; however, the mechanisms by which glia regulate neuronal signaling are still being elucidated. Here, we describe an analysis of how Ca2+ signals within Drosophila astrocyte-like glia regulate excitability in the nervous system. We find that Drosophila astrocytes exhibit robust Ca2+ oscillatory activity manifested by fast, recurrent microdomain Ca2+ fluctuations within processes that infiltrate the synaptic neuropil. Unlike the enhanced neuronal activity and behavioral seizures that were previously observed during manipulations that trigger Ca2+ influx into Drosophila cortex glia, we find that acute induction of astrocyte Ca2+ influx leads to a rapid onset of behavioral paralysis and a suppression of neuronal activity. We observe that Ca2+ influx triggers rapid endocytosis of the GABA transporter (GAT) from astrocyte plasma membranes, suggesting that increased synaptic GABA levels contribute to the neuronal silencing and paralysis. We identify Rab11 as a novel regulator of GAT trafficking that is required for this form of activity regulation. Suppression of Rab11 function strongly offsets the reduction of neuronal activity caused by acute astrocyte Ca2+ influx, likely by inhibiting GAT endocytosis. Our data provide new insights into astrocyte Ca2+ signaling and indicate that distinct glial subtypes in the Drosophila brain can mediate opposing effects on neuronal excitability.
Collapse
|
62
|
Dorot O, Steller H, Segal D, Horowitz M. Past1 Modulates Drosophila Eye Development. PLoS One 2017; 12:e0169639. [PMID: 28060904 PMCID: PMC5218476 DOI: 10.1371/journal.pone.0169639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 12/20/2016] [Indexed: 11/24/2022] Open
Abstract
Endocytosis is a multi-step process involving a large number of proteins, both general factors, such as clathrin and adaptor protein complexes, and unique proteins, which modulate specialized endocytic processes, like the EHD proteins. EHDs are a family of Eps15 Homology Domain containing proteins that consists of four mammalian homologs, one C. elegans, one Drosophila melanogaster and two plants orthologs. These membrane-associated proteins are involved in different steps of endocytic trafficking pathways. We have previously shown that the Drosophila EHD ortholog, PAST1, associates predominantly with the plasma membrane. Mutations in Past1 result in defects in endocytosis, male sterility, temperature sensitivity and premature death of the flies. Also, Past1 genetically interacts with Notch. In the present study, we investigated the role of PAST1 in the developing fly eye. In mutant flies lacking PAST1, abnormal differentiation of photoreceptors R1, R6 and R7 was evident, with partial penetrance. Likewise, five cone cells were present instead of four. Expression of transgenic PAST1 resulted in a dominant negative effect, with a phenotype similar to that of the deletion mutant, and appearance of additional inter-ommatidial pigment cells. Our results strongly suggest a role for PAST1 in differentiation of photoreceptors R1/R6/R7 and cone cells of the fly ommatidia.
Collapse
Affiliation(s)
- Orly Dorot
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Israel
| | - Hermann Steller
- Howard Hughes Medical Institute, Strang Laboratory of Cancer Research, The Rockefeller University, New York, New York, United States of America
| | - Daniel Segal
- Department of Molecular Microbiology and Biotechnology and the Interdisciplinary Sagol School of Neurosciences, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Israel
| | - Mia Horowitz
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Israel
| |
Collapse
|
63
|
Schopf K, Huber A. Membrane protein trafficking in Drosophila photoreceptor cells. Eur J Cell Biol 2016; 96:391-401. [PMID: 27964885 DOI: 10.1016/j.ejcb.2016.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/21/2016] [Accepted: 11/28/2016] [Indexed: 10/20/2022] Open
Abstract
Membrane protein trafficking occurs throughout the lifetime of neurons and includes the initial protein synthesis and anterograde transport to the plasma membrane as well as internalization, degradation, and recycling of plasma membrane proteins. Defects in protein trafficking can result in neuronal degeneration and underlie blinding diseases such as retinitis pigmentosa as well as other neuronal disorders. Drosophila photoreceptor cells have emerged as a model system for identifying the components and mechanisms involved in membrane protein trafficking in neurons. Here we summarize the current knowledge about trafficking of three Drosophila phototransduction proteins, the visual pigment rhodopsin and the two light-activated ion channels TRP (transient receptor potential) and TRPL (TRP-like). Despite some common requirements shared by rhodopsin and TRP, details in the trafficking of these proteins differ considerably, suggesting the existence of several trafficking pathways for these photoreceptor proteins.
Collapse
Affiliation(s)
- Krystina Schopf
- University of Hohenheim, Institute of Physiology, Department of Biosensorics, Stuttgart, Germany
| | - Armin Huber
- University of Hohenheim, Institute of Physiology, Department of Biosensorics, Stuttgart, Germany.
| |
Collapse
|
64
|
Ramaker JM, Cargill RS, Swanson TL, Quirindongo H, Cassar M, Kretzschmar D, Copenhaver PF. Amyloid Precursor Proteins Are Dynamically Trafficked and Processed during Neuronal Development. Front Mol Neurosci 2016; 9:130. [PMID: 27932950 PMCID: PMC5122739 DOI: 10.3389/fnmol.2016.00130] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/10/2016] [Indexed: 01/10/2023] Open
Abstract
Proteolytic processing of the Amyloid Precursor Protein (APP) produces beta-amyloid (Aβ) peptide fragments that accumulate in Alzheimer's Disease (AD), but APP may also regulate multiple aspects of neuronal development, albeit via mechanisms that are not well understood. APP is a member of a family of transmembrane glycoproteins expressed by all higher organisms, including two mammalian orthologs (APLP1 and APLP2) that have complicated investigations into the specific activities of APP. By comparison, insects express only a single APP-related protein (APP-Like, or APPL) that contains the same protein interaction domains identified in APP. However, unlike its mammalian orthologs, APPL is only expressed by neurons, greatly simplifying an analysis of its functions in vivo. Like APP, APPL is processed by secretases to generate a similar array of extracellular and intracellular cleavage fragments, as well as an Aβ-like fragment that can induce neurotoxic responses in the brain. Exploiting the complementary advantages of two insect models (Drosophila melanogaster and Manduca sexta), we have investigated the regulation of APPL trafficking and processing with respect to different aspects of neuronal development. By comparing the behavior of endogenously expressed APPL with fluorescently tagged versions of APPL and APP, we have shown that some full-length protein is consistently trafficked into the most motile regions of developing neurons both in vitro and in vivo. Concurrently, much of the holoprotein is rapidly processed into N- and C-terminal fragments that undergo bi-directional transport within distinct vesicle populations. Unexpectedly, we also discovered that APPL can be transiently sequestered into an amphisome-like compartment in developing neurons, while manipulations targeting APPL cleavage altered their motile behavior in cultured embryos. These data suggest that multiple mechanisms restrict the bioavailability of the holoprotein to regulate APPL-dependent responses within the nervous system. Lastly, targeted expression of our double-tagged constructs (combined with time-lapse imaging) revealed that APP family proteins are subject to complex patterns of trafficking and processing that vary dramatically between different neuronal subtypes. In combination, our results provide a new perspective on how the regulation of APP family proteins can be modulated to accommodate a variety of cell type-specific responses within the embryonic and adult nervous system.
Collapse
Affiliation(s)
- Jenna M Ramaker
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science UniversityPortland, OR, USA; Neuroscience Graduate Program, Oregon Health and Science UniversityPortland, OR, USA
| | - Robert S Cargill
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University Portland, OR, USA
| | - Tracy L Swanson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University Portland, OR, USA
| | - Hanil Quirindongo
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University Portland, OR, USA
| | - Marlène Cassar
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University Portland, OR, USA
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University Portland, OR, USA
| | - Philip F Copenhaver
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University Portland, OR, USA
| |
Collapse
|
65
|
Satoh T, Nakamura Y, Satoh AK. The roles of Syx5 in Golgi morphology and Rhodopsin transport in Drosophila photoreceptors. Biol Open 2016; 5:1420-1430. [PMID: 27591190 PMCID: PMC5087674 DOI: 10.1242/bio.020958] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
SNAREs (SNAP receptors) are the key components of protein complexes that drive membrane fusion. Here, we report the function of a SNARE, Syntaxin 5 (Syx5), in the development of photoreceptors in Drosophila. In wild-type photoreceptors, Syx5 localizes to cis-Golgi, along with cis-Golgi markers: Rab1 and GM130. We observed that Syx5-deficient photoreceptors show notable accumulation of these cis-Golgi markers accompanying drastic accumulation of vesicles between endoplasmic reticulum (ER) and Golgi cisternae. Extensive analysis of Rh1 (rhodopsin 1) trafficking revealed that in Syx5-deficient photoreceptors, Rh1 is exported from the ER with normal kinetics, retained in the cis-Golgi region along with GM130 for a prolonged period, and then subsequently degraded presumably by endoplasmic reticulum-associated protein degradation (ERAD) after retrieval to the ER. Unlike our previous report of Rab6-deficient photoreceptors – where two apical transport pathways are specifically inhibited – vesicle transport pathways to all plasma membrane domains are inhibited in Syx5-deficient photoreceptors, implying that Rab6 and Syx5 are acting in different steps of intra-Golgi transport. These results indicate that Syx5 is crucial for membrane protein transport, presumably during ER-derived vesicle fusion to form cis-Golgi cisternae. Summary: Unlike Rab6-deficiency which affects only apical transport pathways, Syx5-deficiency inhibits all of polarized transport pathways, implying that these two genes are working in distinct stages of intra-Golgi transport.
Collapse
Affiliation(s)
- Takunori Satoh
- Division of Life Science, Graduate School of Integral Arts and Science, Hiroshima University, 1-7-1, Kagamiyama, Higashi-hiroshima 739-8521, Japan
| | - Yuri Nakamura
- Division of Life Science, Graduate School of Integral Arts and Science, Hiroshima University, 1-7-1, Kagamiyama, Higashi-hiroshima 739-8521, Japan
| | - Akiko K Satoh
- Division of Life Science, Graduate School of Integral Arts and Science, Hiroshima University, 1-7-1, Kagamiyama, Higashi-hiroshima 739-8521, Japan
| |
Collapse
|
66
|
Chaharbakhshi E, Jemc JC. Broad-complex, tramtrack, and bric-à-brac (BTB) proteins: Critical regulators of development. Genesis 2016; 54:505-518. [DOI: 10.1002/dvg.22964] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/08/2016] [Accepted: 08/11/2016] [Indexed: 01/21/2023]
Affiliation(s)
- Edwin Chaharbakhshi
- Department of Biology; Loyola University Chicago; Chicago IL
- Stritch School of Medicine; Loyola University Chicago; Maywood IL
| | | |
Collapse
|
67
|
Deshpande M, Feiger Z, Shilton AK, Luo CC, Silverman E, Rodal AA. Role of BMP receptor traffic in synaptic growth defects in an ALS model. Mol Biol Cell 2016; 27:2898-910. [PMID: 27535427 PMCID: PMC5042577 DOI: 10.1091/mbc.e16-07-0519] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 08/03/2016] [Indexed: 12/12/2022] Open
Abstract
In a Drosophila model of ALS, neuronal defects are associated with altered endosomal traffic of growth factor receptors and loss of growth-promoting signals. Manipulation of an endosomal recycling pathway suppresses these neuronal defects. The findings suggest that rerouting membrane traffic could be therapeutic in ALS. TAR DNA-binding protein 43 (TDP-43) is genetically and functionally linked to amyotrophic lateral sclerosis (ALS) and regulates transcription, splicing, and transport of thousands of RNA targets that function in diverse cellular pathways. In ALS, pathologically altered TDP-43 is believed to lead to disease by toxic gain-of-function effects on RNA metabolism, as well as by sequestering endogenous TDP-43 and causing its loss of function. However, it is unclear which of the numerous cellular processes disrupted downstream of TDP-43 dysfunction lead to neurodegeneration. Here we found that both loss and gain of function of TDP-43 in Drosophila cause a reduction of synaptic growth–promoting bone morphogenic protein (BMP) signaling at the neuromuscular junction (NMJ). Further, we observed a shift of BMP receptors from early to recycling endosomes and increased mobility of BMP receptor–containing compartments at the NMJ. Inhibition of the recycling endosome GTPase Rab11 partially rescued TDP-43–induced defects in BMP receptor dynamics and distribution and suppressed BMP signaling, synaptic growth, and larval crawling defects. Our results indicate that defects in receptor traffic lead to neuronal dysfunction downstream of TDP-43 misregulation and that rerouting receptor traffic may be a viable strategy for rescuing neurological impairment.
Collapse
Affiliation(s)
| | - Zachary Feiger
- Department of Biology, Brandeis University, Waltham, MA 02453
| | | | - Christina C Luo
- Department of Biology, Brandeis University, Waltham, MA 02453
| | - Ethan Silverman
- Department of Biology, Brandeis University, Waltham, MA 02453
| | - Avital A Rodal
- Department of Biology, Brandeis University, Waltham, MA 02453
| |
Collapse
|
68
|
Small GTPases Rab8a and Rab11a Are Dispensable for Rhodopsin Transport in Mouse Photoreceptors. PLoS One 2016; 11:e0161236. [PMID: 27529348 PMCID: PMC4987053 DOI: 10.1371/journal.pone.0161236] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 08/02/2016] [Indexed: 01/01/2023] Open
Abstract
Rab11a and Rab8a are ubiquitous small GTPases shown as required for rhodopsin transport in Xenopus laevis and zebrafish photoreceptors by dominant negative (dn) disruption of function. Here, we generated retina-specific Rab11a (retRab11a) and Rab8a (retRab8a) single and double knockout mice to explore the consequences in mouse photoreceptors. Rhodopsin and other outer segment (OS) membrane proteins targeted correctly to OS and electroretinogram (ERG) responses in all three mutant mouse lines were indistinguishable from wild-type (WT). Further, AAV (adeno-associated virus)-mediated expression of dnRab11b in retRab11a-/- retina, or expression of dnRab8b in retRab8a-/- retina did not cause OS protein mislocalization. Finally, a retRab8a-/- retina injected at one month of age with AAVs expressing dnRab11a, dnRab11b, dnRab8b, and dnRab10 (four dn viruses on Rab8a-/- background) and harvested three months later exhibited normal OS protein localization. In contrast to results obtained with dnRab GTPases in Xenopus and zebrafish, mouse Rab11a and Rab8a are dispensable for proper rhodopsin and outer segment membrane protein targeting. Absence of phenotype after expression of four dn Rab GTPases in a Rab8a-/- retina suggests that Rab8b and Rab11b paralogs maybe dispensable as well. Our data thus demonstrate significant interspecies variation in photoreceptor membrane protein and rhodopsin trafficking.
Collapse
|
69
|
Román-Fernández A, Bryant DM. Complex Polarity: Building Multicellular Tissues Through Apical Membrane Traffic. Traffic 2016; 17:1244-1261. [PMID: 27281121 DOI: 10.1111/tra.12417] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 06/06/2016] [Accepted: 06/06/2016] [Indexed: 12/20/2022]
Abstract
The formation of distinct subdomains of the cell surface is crucial for multicellular organism development. The most striking example of this is apical-basal polarization. What is much less appreciated is that underpinning an asymmetric cell surface is an equally dramatic intracellular endosome rearrangement. Here, we review the interplay between classical cell polarity proteins and membrane trafficking pathways, and discuss how this marriage gives rise to cell polarization. We focus on those mechanisms that regulate apical polarization, as this is providing a number of insights into how membrane traffic and polarity are regulated at the tissue level.
Collapse
Affiliation(s)
- Alvaro Román-Fernández
- Cancer Research UK Beatson Institute, Switchback Road, Glasgow, G61 1BD, UK.,Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - David M Bryant
- Cancer Research UK Beatson Institute, Switchback Road, Glasgow, G61 1BD, UK.,Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| |
Collapse
|
70
|
Sorvina A, Shandala T, Brooks DA. Drosophila Pkaap regulates Rab4/Rab11-dependent traffic and Rab11 exocytosis of innate immune cargo. Biol Open 2016; 5:678-88. [PMID: 27190105 PMCID: PMC4920182 DOI: 10.1242/bio.016642] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The secretion of immune-mediators is a critical step in the host innate immune response to pathogen invasion, and Rab GTPases have an important role in the regulation of this process. Rab4/Rab11 recycling endosomes are involved in the sorting of immune-mediators into specialist Rab11 vesicles that can traffic this cargo to the plasma membrane; however, how this sequential delivery process is regulated has yet to be fully defined. Here, we report that Drosophila Pkaap, an orthologue of the human dual-specific A-kinase-anchoring protein 2 or D-AKAP2 (also called AKAP10), appeared to have a nucleotide-dependent localisation to Rab4 and Rab11 endosomes. RNAi silencing of pkaap altered Rab4/Rab11 recycling endosome morphology, suggesting that Pkaap functions in cargo sorting and delivery in the secretory pathway. The depletion of pkaap also had a direct effect on Rab11 vesicle exocytosis and the secretion of the antimicrobial peptide Drosomycin at the plasma membrane. We propose that Pkaap has a dual role in antimicrobial peptide traffic and exocytosis, making it an essential component for the secretion of inflammatory mediators and the defence of the host against pathogens.
Collapse
Affiliation(s)
- Alexandra Sorvina
- Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Tetyana Shandala
- Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Douglas A Brooks
- Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia 5001, Australia
| |
Collapse
|
71
|
Abstract
Selective membrane transport pathways are essential for cells in situ to construct and maintain a polarized structure comprising multiple plasma membrane domains, which is essential for their specific cellular functions. Genetic screening in Drosophila photoreceptors harboring multiple plasma membrane domains enables the identification of genes involved in polarized transport pathways. Our genome-wide high-throughput screening identified a Rab6-null mutant with a rare phenotype characterized by a loss of 2 apical transport pathways with an intact basolateral transport. Although the functions of Rab6 in the Golgi apparatus are well known, its function in polarized transport is unexpected. The mutant phenotype and localization of Rab6 strongly indicate that Rab6 regulates transport between the trans-Golgi network (TGN) and recycling endosomes (REs): basolateral cargos are segregated at the TGN before Rab6 functions, but cargos going to multiple apical domains are sorted at REs. Both the medial-Golgi resident protein Metallophosphoesterase (MPPE) and the TGN marker GalT::CFP exhibit diffused co-localized distributions in Rab6-deficient cells, suggesting they are trapped in the retrograde transport vesicles returning to trans-Golgi cisternae. Hence, we propose that Rab6 regulates the fusion of retrograde transport vesicles containing medial, trans-Golgi resident proteins to the Golgi cisternae, which causes Golgi maturation to REs.
Collapse
Affiliation(s)
- Takunori Satoh
- a Division of Life Science, Graduate School of Integral Arts and Science, Hiroshima University , Higashi-Hiroshima , Japan
| | - Yuri Nakamura
- a Division of Life Science, Graduate School of Integral Arts and Science, Hiroshima University , Higashi-Hiroshima , Japan
| | - Akiko K Satoh
- a Division of Life Science, Graduate School of Integral Arts and Science, Hiroshima University , Higashi-Hiroshima , Japan
| |
Collapse
|
72
|
Mavor LM, Miao H, Zuo Z, Holly RM, Xie Y, Loerke D, Blankenship JT. Rab8 directs furrow ingression and membrane addition during epithelial formation in Drosophila melanogaster. Development 2016; 143:892-903. [PMID: 26839362 PMCID: PMC4813336 DOI: 10.1242/dev.128876] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 01/25/2016] [Indexed: 01/31/2023]
Abstract
One of the most fundamental changes in cell morphology is the ingression of a plasma membrane furrow. The Drosophila embryo undergoes several cycles of rapid furrow ingression during early development that culminate in the formation of an epithelial sheet. Previous studies have demonstrated the requirement for intracellular trafficking pathways in furrow ingression; however, the pathways that link compartmental behaviors with cortical furrow ingression events are unclear. Here, we show that Rab8 has striking dynamic behaviors in vivo. As furrows ingress, cytoplasmic Rab8 puncta are depleted and Rab8 accumulates at the plasma membrane in a location that coincides with known regions of directed membrane addition. We additionally use CRISPR/Cas9 technology to N-terminally tag Rab8, which is then used to address endogenous localization and function. Endogenous Rab8 displays partial coincidence with Rab11 and the Golgi, and this colocalization is enriched during the fast phase of cellularization. When Rab8 function is disrupted, furrow formation in the early embryo is completely abolished. We also demonstrate that Rab8 behaviors require the function of the exocyst complex subunit Sec5 as well as the recycling endosome protein Rab11. Active, GTP-locked Rab8 is primarily associated with dynamic membrane compartments and the plasma membrane, whereas GDP-locked Rab8 forms large cytoplasmic aggregates. These studies suggest a model in which active Rab8 populations direct furrow ingression by guiding the targeted delivery of cytoplasmic membrane stores to the cell surface through interactions with the exocyst tethering complex.
Collapse
Affiliation(s)
- Lauren M Mavor
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | - Hui Miao
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | - Zhongyuan Zuo
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | - Ryan M Holly
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | - Yi Xie
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | - Dinah Loerke
- Department of Physics, University of Denver, Denver, CO 80208, USA
| | - J Todd Blankenship
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| |
Collapse
|
73
|
Iwanami N, Nakamura Y, Satoh T, Liu Z, Satoh AK. Rab6 Is Required for Multiple Apical Transport Pathways but Not the Basolateral Transport Pathway in Drosophila Photoreceptors. PLoS Genet 2016; 12:e1005828. [PMID: 26890939 PMCID: PMC4758697 DOI: 10.1371/journal.pgen.1005828] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 01/05/2016] [Indexed: 11/30/2022] Open
Abstract
Polarized membrane trafficking is essential for the construction and maintenance of multiple plasma membrane domains of cells. Highly polarized Drosophila photoreceptors are an excellent model for studying polarized transport. A single cross-section of Drosophila retina contains many photoreceptors with 3 clearly differentiated plasma membrane domains: a rhabdomere, stalk, and basolateral membrane. Genome-wide high-throughput ethyl methanesulfonate screening followed by precise immunohistochemical analysis identified a mutant with a rare phenotype characterized by a loss of 2 apical transport pathways with normal basolateral transport. Rapid gene identification using whole-genome resequencing and single nucleotide polymorphism mapping identified a nonsense mutation of Rab6 responsible for the apical-specific transport deficiency. Detailed analysis of the trafficking of a major rhabdomere protein Rh1 using blue light-induced chromophore supply identified Rab6 as essential for Rh1 to exit the Golgi units. Rab6 is mostly distributed from the trans-Golgi network to a Golgi-associated Rab11-positive compartment that likely recycles endosomes or transport vesicles going to recycling endosomes. Furthermore, the Rab6 effector, Rich, is required for Rab6 recruitment in the trans-Golgi network. Moreover, a Rich null mutation phenocopies the Rab6 null mutant, indicating that Rich functions as a guanine nucleotide exchange factor for Rab6. The results collectively indicate that Rab6 and Rich are essential for the trans-Golgi network–recycling endosome transport of cargoes destined for 2 apical domains. However, basolateral cargos are sorted and exported from the trans-Golgi network in a Rab6-independent manner. Cells in animal bodies have multiple plasma membrane domains; this polarized characteristic of cells is essential for their specific functions. Selective membrane transport pathways play key roles in the construction and maintenance of polarized structures. Drosophila photoreceptors with multiple plasma membrane domains are an excellent model of polarized transport. We performed genetic screening and identified a Rab6 null mutant with a rare phenotype characterized by a loss of 2 apical transport pathways with normal basolateral transport. Although Rab6 functions in the Golgi are well known, its function in polarized transport was unexpected. Here, we found that Rab6 and its effector, Rich, are required for multiple apical transport pathways but not the basolateral transport pathway. Our findings strongly indicate that the membrane proteins delivered to multiple polarized domains are not sorted simultaneously: basolateral cargos are segregated before the Rab6-dependent process, and cargos going to multiple apical domains are sorted after Rab6-dependent transport from the trans-Golgi network to the Golgi-associated Rab11-positive compartment, which presumably recycles endosomes. Our finding of the function of Rab6 in polarized transport will elucidate the molecular mechanisms of polarized transport.
Collapse
Affiliation(s)
- Nozomi Iwanami
- Division of Life Science, Graduate School of Integral Arts and Science, Hiroshima University, Higashi-Hiroshima, Japan
| | - Yuri Nakamura
- Division of Life Science, Graduate School of Integral Arts and Science, Hiroshima University, Higashi-Hiroshima, Japan
| | - Takunori Satoh
- Division of Life Science, Graduate School of Integral Arts and Science, Hiroshima University, Higashi-Hiroshima, Japan
| | - Ziguang Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Akiko K. Satoh
- Division of Life Science, Graduate School of Integral Arts and Science, Hiroshima University, Higashi-Hiroshima, Japan
- * E-mail:
| |
Collapse
|
74
|
Xu Z, Chikka MR, Xia H, Ready DF. Ire1 supports normal ER differentiation in developing Drosophila photoreceptors. J Cell Sci 2016; 129:921-9. [PMID: 26787744 PMCID: PMC4813318 DOI: 10.1242/jcs.180406] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 01/12/2016] [Indexed: 01/09/2023] Open
Abstract
The endoplasmic reticulum (ER) serves virtually all aspects of cell physiology and, by pathways that are incompletely understood, is dynamically remodeled to meet changing cell needs. Inositol-requiring enzyme 1 (Ire1), a conserved core protein of the unfolded protein response (UPR), participates in ER remodeling and is particularly required during the differentiation of cells devoted to intense secretory activity, so-called 'professional' secretory cells. Here, we characterize the role of Ire1 in ER differentiation in the developing Drosophila compound eye photoreceptors (R cells). As part of normal development, R cells take a turn as professional secretory cells with a massive secretory effort that builds the photosensitive membrane organelle, the rhabdomere. We find rough ER sheets proliferate as rhabdomere biogenesis culminates, and Ire1 is required for normal ER differentiation. Ire1 is active early in R cell development and is required in anticipation of peak biosynthesis. Without Ire1, the amount of rough ER sheets is strongly reduced and the extensive cortical ER network at the rhabdomere base, the subrhabdomere cisterna (SRC), fails. Instead, ER proliferates in persistent and ribosome-poor tubular tangles. A phase of Ire1 activity early in R cell development thus shapes dynamic ER.
Collapse
Affiliation(s)
- Zuyuan Xu
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | | - Hongai Xia
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Donald F Ready
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
75
|
The GTP- and Phospholipid-Binding Protein TTD14 Regulates Trafficking of the TRPL Ion Channel in Drosophila Photoreceptor Cells. PLoS Genet 2015; 11:e1005578. [PMID: 26509977 PMCID: PMC4624897 DOI: 10.1371/journal.pgen.1005578] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 09/15/2015] [Indexed: 01/04/2023] Open
Abstract
Recycling of signaling proteins is a common phenomenon in diverse signaling pathways. In photoreceptors of Drosophila, light absorption by rhodopsin triggers a phospholipase Cβ-mediated opening of the ion channels transient receptor potential (TRP) and TRP-like (TRPL) and generates the visual response. The signaling proteins are located in a plasma membrane compartment called rhabdomere. The major rhodopsin (Rh1) and TRP are predominantly localized in the rhabdomere in light and darkness. In contrast, TRPL translocates between the rhabdomeral plasma membrane in the dark and a storage compartment in the cell body in the light, from where it can be recycled to the plasma membrane upon subsequent dark adaptation. Here, we identified the gene mutated in trpl translocation defective 14 (ttd14), which is required for both TRPL internalization from the rhabdomere in the light and recycling of TRPL back to the rhabdomere in the dark. TTD14 is highly conserved in invertebrates and binds GTP in vitro. The ttd14 mutation alters a conserved proline residue (P75L) in the GTP-binding domain and abolishes binding to GTP. This indicates that GTP binding is essential for TTD14 function. TTD14 is a cytosolic protein and binds to PtdIns(3)P, a lipid enriched in early endosome membranes, and to phosphatidic acid. In contrast to TRPL, rhabdomeral localization of the membrane proteins Rh1 and TRP is not affected in the ttd14P75L mutant. The ttd14P75L mutation results in Rh1-independent photoreceptor degeneration and larval lethality suggesting that other processes are also affected by the ttd14P75L mutation. In conclusion, TTD14 is a novel regulator of TRPL trafficking, involved in internalization and subsequent sorting of TRPL into the recycling pathway that enables this ion channel to return to the plasma membrane. Protein trafficking in neurons occurs throughout the lifetime of a cell and includes the internalization and redistribution of plasma membrane proteins. Regulated protein trafficking controls the equipment of the plasma membrane with receptors and ion channels and thereby attenuates or enhances neuronal function. Defects in recycling of plasma membrane proteins can cause detrimental neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease and Down´s syndrome. In Drosophila photoreceptors, the TRPL ion channel, together with the TRP channel, mediates vision and light-dependently shuttles between an endomembrane storage compartment and the apical plasma membrane. Here, we report the identification of a mutation in the ttd14 gene that inhibits TRPL-trafficking in both directions and also results in photoreceptor degeneration. The TTD14 protein contains a region with weak homology to a PX-domain, which is also found in proteins that sort cargo in the endosome and enable protein recycling. We characterize TTD14 as a new regulator of photoreceptor maintenance and ion channel trafficking that binds to GTP and PtdIns(3)P, a phospholipid enriched in early endosomes.
Collapse
|
76
|
TRP and Rhodopsin Transport Depends on Dual XPORT ER Chaperones Encoded by an Operon. Cell Rep 2015; 13:573-584. [PMID: 26456832 DOI: 10.1016/j.celrep.2015.09.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/30/2015] [Accepted: 09/04/2015] [Indexed: 11/24/2022] Open
Abstract
TRP channels and G protein-coupled receptors (GPCRs) play critical roles in sensory reception. However, the identities of the chaperones that assist GPCRs in translocating from the endoplasmic reticulum (ER) are limited, and TRP ER chaperones are virtually unknown. The one exception for TRPs is Drosophila XPORT. Here, we show that the xport locus is bicistronic and encodes unrelated transmembrane proteins, which enable the signaling proteins that initiate and culminate phototransduction, rhodopsin 1 (Rh1) and TRP, to traffic to the plasma membrane. XPORT-A and XPORT-B are ER proteins, and loss of either has a profound impact on TRP and Rh1 targeting to the light-sensing compartment of photoreceptor cells. XPORT-B complexed in vivo with the Drosophila homolog of the mammalian HSP70 protein, GRP78/BiP, which, in turn, associated with Rh1. Our work highlights a coordinated network of chaperones required for the biosynthesis of the TRP channel and rhodopsin in Drosophila photoreceptor cells.
Collapse
|
77
|
Schleede J, Blair SS. The Gyc76C Receptor Guanylyl Cyclase and the Foraging cGMP-Dependent Kinase Regulate Extracellular Matrix Organization and BMP Signaling in the Developing Wing of Drosophila melanogaster. PLoS Genet 2015; 11:e1005576. [PMID: 26440503 PMCID: PMC4595086 DOI: 10.1371/journal.pgen.1005576] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/16/2015] [Indexed: 12/30/2022] Open
Abstract
The developing crossveins of the wing of Drosophila melanogaster are specified by long-range BMP signaling and are especially sensitive to loss of extracellular modulators of BMP signaling such as the Chordin homolog Short gastrulation (Sog). However, the role of the extracellular matrix in BMP signaling and Sog activity in the crossveins has been poorly explored. Using a genetic mosaic screen for mutations that disrupt BMP signaling and posterior crossvein development, we identify Gyc76C, a member of the receptor guanylyl cyclase family that includes mammalian natriuretic peptide receptors. We show that Gyc76C and the soluble cGMP-dependent kinase Foraging, likely linked by cGMP, are necessary for normal refinement and maintenance of long-range BMP signaling in the posterior crossvein. This does not occur through cell-autonomous crosstalk between cGMP and BMP signal transduction, but likely through altered extracellular activity of Sog. We identify a novel pathway leading from Gyc76C to the organization of the wing extracellular matrix by matrix metalloproteinases, and show that both the extracellular matrix and BMP signaling effects are largely mediated by changes in the activity of matrix metalloproteinases. We discuss parallels and differences between this pathway and other examples of cGMP activity in both Drosophila melanogaster and mammalian cells and tissues. Signaling between cells regulates many processes, including the choices cells make between different fates during development and regeneration, and misregulation of such signaling underlies many human pathologies. To understand how such signals control developmental decisions, it is necessary to elucidate both how cells regulate and respond to different levels of signaling, and how different types of signals combine and regulate each other. We have used genetic screening in the fruitfly Drosophila melanogaster to identify mutations that reduce or eliminate signals carried by Bone Morphogenetic Proteins (BMPs), and show that BMP signaling is sensitive Gyc76C, a peptide receptor that stimulates the production of cGMP in cells. We identify downstream intracellular effectors of this cGMP activity, but provide evidence that the effects on the BMP pathway are not mediated at the intracellular level, but rather through cGMP’s effects upon the extracellular matrix and matrix-remodeling proteinases, which in turn affects the activity of extracellular BMP-binding proteins. We discuss differences and parallels with other examples of cGMP activity in Drosophila melanogaster and mammals.
Collapse
Affiliation(s)
- Justin Schleede
- Department of Zoology, University of Wisconsin, Madison, Wisconsin, United States of America
- Genetics Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Seth S. Blair
- Department of Zoology, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
78
|
Bader CA, Shandala T, Ng YS, Johnson IRD, Brooks DA. Atg9 is required for intraluminal vesicles in amphisomes and autolysosomes. Biol Open 2015; 4:1345-55. [PMID: 26353861 PMCID: PMC4728360 DOI: 10.1242/bio.013979] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Autophagy is an intracellular recycling and degradation process, which is important for energy metabolism, lipid metabolism, physiological stress response and organism development. During Drosophila development, autophagy is up-regulated in fat body and midgut cells, to control metabolic function and to enable tissue remodelling. Atg9 is the only transmembrane protein involved in the core autophagy machinery and is thought to have a role in autophagosome formation. During Drosophila development, Atg9 co-located with Atg8 autophagosomes, Rab11 endosomes and Lamp1 endosomes-lysosomes. RNAi silencing of Atg9 reduced both the number and the size of autophagosomes during development and caused morphological changes to amphisomes/autolysosomes. In control cells there was compartmentalised acidification corresponding to intraluminal Rab11/Lamp-1 vesicles, but in Atg9 depleted cells there were no intraluminal vesicles and the acidification was not compartmentalised. We concluded that Atg9 is required to form intraluminal vesicles and for localised acidification within amphisomes/autolysosomes, and consequently when depleted, reduced the capacity to degrade and remodel gut tissue during development. Summary: The disappearance of intraluminal vesicles in amphisomes/autolysosomes upon Atg9 depletion suggests that Atg9 has a specific role in intraluminal vesicle formation in autophagic compartments.
Collapse
Affiliation(s)
- C A Bader
- Mechanisms in Cell Biology and Diseases Research Group, School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia 5001, Australia
| | - T Shandala
- Mechanisms in Cell Biology and Diseases Research Group, School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Y S Ng
- Mechanisms in Cell Biology and Diseases Research Group, School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia 5001, Australia
| | - I R D Johnson
- Mechanisms in Cell Biology and Diseases Research Group, School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia 5001, Australia
| | - D A Brooks
- Mechanisms in Cell Biology and Diseases Research Group, School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia 5001, Australia
| |
Collapse
|
79
|
Zang Y, Wan M, Liu M, Ke H, Ma S, Liu LP, Ni JQ, Pastor-Pareja JC. Plasma membrane overgrowth causes fibrotic collagen accumulation and immune activation in Drosophila adipocytes. eLife 2015; 4:e07187. [PMID: 26090908 PMCID: PMC4490375 DOI: 10.7554/elife.07187] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/18/2015] [Indexed: 12/13/2022] Open
Abstract
Many chronic diseases are associated with fibrotic deposition of Collagen and other matrix proteins. Little is known about the factors that determine preferential onset of fibrosis in particular tissues. Here we show that plasma membrane (PM) overgrowth causes pericellular Collagen accumulation in Drosophila adipocytes. We found that loss of Dynamin and other endocytic components causes pericellular trapping of outgoing Collagen IV due to dramatic cortex expansion when endocytic removal of PM is prevented. Deposits also form in the absence of negative Toll immune regulator Cactus, excess PM being caused in this case by increased secretion. Finally, we show that trimeric Collagen accumulation, downstream of Toll or endocytic defects, activates a tissue damage response. Our work indicates that traffic imbalances and PM topology may contribute to fibrosis. It also places fibrotic deposits both downstream and upstream of immune signaling, consistent with the chronic character of fibrotic diseases.
Collapse
Affiliation(s)
- Yiran Zang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Ming Wan
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Min Liu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Hongmei Ke
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Shuangchun Ma
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Lu-Ping Liu
- Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, China
| | - Jian-Quan Ni
- Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, China
| | | |
Collapse
|
80
|
Wan D, Zhang ZC, Zhang X, Li Q, Han J. X chromosome-linked intellectual disability protein PQBP1 associates with and regulates the translation of specific mRNAs. Hum Mol Genet 2015; 24:4599-614. [DOI: 10.1093/hmg/ddv191] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/19/2015] [Indexed: 01/08/2023] Open
|
81
|
PI3K-C2α: One enzyme for two products coupling vesicle trafficking and signal transduction. FEBS Lett 2015; 589:1552-8. [DOI: 10.1016/j.febslet.2015.05.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/04/2015] [Accepted: 05/05/2015] [Indexed: 12/20/2022]
|
82
|
The motor function of Drosophila melanogaster myosin-5 is activated by calcium and cargo-binding protein dRab11. Biochem J 2015; 469:135-44. [PMID: 25940004 DOI: 10.1042/bj20141330] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 05/05/2015] [Indexed: 12/14/2022]
Abstract
In the Drosophila melanogaster compound eye, myosin-5 (DmM5) plays two distinct roles in response to light stimulation: transport of pigment granules to the rhabdomere base to decrease light exposure and transport of rhodopsin-bearing vesicles to the rhabdomere base to compensate for the rhodopsin loss during light exposure. However, little is known of how the motor function of DmM5 is regulated at the molecular level. In the present study, we overexpressed DmM5 in Sf9 insect cells and investigated its regulation using purified proteins. We found that the actin-activated ATPase activity of DmM5 is significantly lower than that of the truncated DmM5 having the C-terminal globular tail domain (GTD) deleted, indicating that the GTD is the inhibitory domain. The actin-activated ATPase activity of DmM5 is significantly activated by micromolar levels of calcium. DmM5 associates with pigment granules and rhodopsin-bearing vesicles through cargo-binding proteins Lightoid (Ltd) and dRab11 respectively. We found that GTP-bound dRab11, but not Ltd, significantly activates DmM5 actin-activated ATPase activity. Moreover, we identified Gln(1689) in the GTD as the critical residue for the interaction with dRab11 and activation of DmM5 motor function by dRab11. Based on those results, we propose that DmM5-dependent transport of pigment granules is directly activated by light-induced calcium influx and the DmM5-dependent transport of rhodopsin-bearing vesicle is activated by active GTP-bound dRab11, whose formation is stimulated by light-induced calcium influx.
Collapse
|
83
|
Bark beetle controls epithelial morphogenesis by septate junction maturation in Drosophila. Dev Biol 2015; 400:237-47. [DOI: 10.1016/j.ydbio.2015.02.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/04/2015] [Accepted: 02/11/2015] [Indexed: 12/18/2022]
|
84
|
Kelley M, Yochem J, Krieg M, Calixto A, Heiman MG, Kuzmanov A, Meli V, Chalfie M, Goodman MB, Shaham S, Frand A, Fay DS. FBN-1, a fibrillin-related protein, is required for resistance of the epidermis to mechanical deformation during C. elegans embryogenesis. eLife 2015; 4. [PMID: 25798732 PMCID: PMC4395870 DOI: 10.7554/elife.06565] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/20/2015] [Indexed: 12/19/2022] Open
Abstract
During development, biomechanical forces contour the body and provide shape to internal organs. Using genetic and molecular approaches in combination with a FRET-based tension sensor, we characterized a pulling force exerted by the elongating pharynx (foregut) on the anterior epidermis during C. elegans embryogenesis. Resistance of the epidermis to this force and to actomyosin-based circumferential constricting forces is mediated by FBN-1, a ZP domain protein related to vertebrate fibrillins. fbn-1 was required specifically within the epidermis and FBN-1 was expressed in epidermal cells and secreted to the apical surface as a putative component of the embryonic sheath. Tiling array studies indicated that fbn-1 mRNA processing requires the conserved alternative splicing factor MEC-8/RBPMS. The conserved SYM-3/FAM102A and SYM-4/WDR44 proteins, which are linked to protein trafficking, function as additional components of this network. Our studies demonstrate the importance of the apical extracellular matrix in preventing mechanical deformation of the epidermis during development. DOI:http://dx.doi.org/10.7554/eLife.06565.001 For an animal embryo to develop, its cells must organize themselves into tissues and organs. For example, skin and the lining of internal organs—such as the lungs and gut—are made from cells called epithelial cells, which are tightly linked to form flat sheets. In a microscopic worm called Caenorhabditis elegans, the outermost layer of epithelial cells (called the epidermis) forms over the surface of the embryo early on in embryonic development. Shortly afterwards, the embryonic epidermis experiences powerful contractions along the surface of the embryo. The force generated by these contractions converts the embryo from an oval shape to a roughly cylindrical form. These contractions also squeeze the internal tissues and organs, which correspondingly elongate along with the epidermis. It has been known for decades that such ‘mechanical’ forces are important for the normal development of embryos. However, it remains poorly understood how these forces generate tissues and organs of the proper shape—partly because it is difficult to measure forces in living embryos. It is also not clear how the mechanical properties of specific tissues are controlled. Now, Kelley, Yochem, Krieg et al. have analyzed the development of C. elegans' embryos and discovered a novel mechanical interplay between the feeding organ (called the pharynx) and the worm's epidermis. The experiments involved studying several mutant worms that perturb epidermal contractions and disrupt the attachment of the pharynx to the epidermis. These studies suggested that the pharynx exerts a strong inward pulling force on the epidermis during development. Using recently developed methods, Kelley, Yochem, Krieg et al. then measured mechanical forces within intact worm embryos and demonstrated that greater forces were experienced in cells that were being pulled by the pharynx. Kelley, Yochem, Krieg et al. further analyzed how the epidermis normally resists this pulling force from the pharynx and implicated a protein called FBN-1. This worm protein is structurally related to a human protein that is affected in people with a disorder called Marfan Syndrome. Worm embryos without the FBN-1 protein become severely deformed because they are unable to withstand mechanical forces at the epidermis. FBN-1 is normally synthesized and then transported to the outside of the worm embryo by epidermal cells, where it is thought to assemble into a meshwork of long fibers. This provides a strong scaffold that attaches to the epidermis to prevent the epidermis from undergoing excessive deformation while it experiences mechanical forces. The work of Kelley, Yochem, Krieg et al. provides an opportunity to understand how FBN-1 and other fiber-forming proteins are produced and transported to the cell surface. Moreover, these findings may have implications for human diseases and birth defects that result from an inability of tissues to respond appropriately to mechanical forces. DOI:http://dx.doi.org/10.7554/eLife.06565.002
Collapse
Affiliation(s)
- Melissa Kelley
- Department of Molecular Biology, University of Wyoming, Laramie, United States
| | - John Yochem
- Department of Molecular Biology, University of Wyoming, Laramie, United States
| | - Michael Krieg
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| | - Andrea Calixto
- Department of Biological Sciences, Columbia University, New York, United States
| | - Maxwell G Heiman
- Department of Genetics, Harvard Medical School, Boston Children's Hospital, Boston, United States
| | - Aleksandra Kuzmanov
- Department of Molecular Biology, University of Wyoming, Laramie, United States
| | - Vijaykumar Meli
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, United States
| | - Martin Chalfie
- Department of Biological Sciences, Columbia University, New York, United States
| | - Miriam B Goodman
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, New York, United States
| | - Alison Frand
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, United States
| | - David S Fay
- Department of Molecular Biology, University of Wyoming, Laramie, United States
| |
Collapse
|
85
|
Legent K, Liu HH, Treisman JE. Drosophila Vps4 promotes Epidermal growth factor receptor signaling independently of its role in receptor degradation. Development 2015; 142:1480-91. [PMID: 25790850 DOI: 10.1242/dev.117960] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 02/20/2015] [Indexed: 12/12/2022]
Abstract
Endocytic trafficking of signaling receptors is an important mechanism for limiting signal duration. Components of the Endosomal Sorting Complexes Required for Transport (ESCRT), which target ubiquitylated receptors to intra-lumenal vesicles (ILVs) of multivesicular bodies, are thought to terminate signaling by the epidermal growth factor receptor (EGFR) and direct it for lysosomal degradation. In a genetic screen for mutations that affect Drosophila eye development, we identified an allele of Vacuolar protein sorting 4 (Vps4), which encodes an AAA ATPase that interacts with the ESCRT-III complex to drive the final step of ILV formation. Photoreceptors are largely absent from Vps4 mutant clones in the eye disc, and even when cell death is genetically prevented, the mutant R8 photoreceptors that develop fail to recruit surrounding cells to differentiate as R1-R7 photoreceptors. This recruitment requires EGFR signaling, suggesting that loss of Vps4 disrupts the EGFR pathway. In imaginal disc cells mutant for Vps4, EGFR and other receptors accumulate in endosomes and EGFR target genes are not expressed; epistasis experiments place the function of Vps4 at the level of the receptor. Surprisingly, Vps4 is required for EGFR signaling even in the absence of Shibire, the Dynamin that internalizes EGFR from the plasma membrane. In ovarian follicle cells, in contrast, Vps4 does not affect EGFR signaling, although it is still essential for receptor degradation. Taken together, these findings indicate that Vps4 can promote EGFR activity through an endocytosis-independent mechanism.
Collapse
Affiliation(s)
- Kevin Legent
- Kimmel Center for Biology and Medicine of the Skirball Institute and Department of Cell Biology, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Hui Hua Liu
- Kimmel Center for Biology and Medicine of the Skirball Institute and Department of Cell Biology, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Jessica E Treisman
- Kimmel Center for Biology and Medicine of the Skirball Institute and Department of Cell Biology, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA
| |
Collapse
|
86
|
Le Droguen PM, Claret S, Guichet A, Brodu V. Microtubule-dependent apical restriction of recycling endosomes sustains adherens junctions during morphogenesis of the Drosophila tracheal system. Development 2015; 142:363-74. [PMID: 25564624 DOI: 10.1242/dev.113472] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Epithelial remodelling is an essential mechanism for organogenesis, during which cells change shape and position while maintaining contact with each other. Adherens junctions (AJs) mediate stable intercellular cohesion but must be actively reorganised to allow morphogenesis. Vesicle trafficking and the microtubule (MT) cytoskeleton contribute to regulating AJs but their interrelationship remains elusive. We carried out a detailed analysis of the role of MTs in cell remodelling during formation of the tracheal system in the Drosophila embryo. Induction of MT depolymerisation specifically in tracheal cells shows that MTs are essential during a specific time frame of tracheal cell elongation while the branch extends. In the absence of MTs, one tracheal cell per branch overelongates, ultimately leading to branch break. Three-dimensional quantifications revealed that MTs are crucial to sustain E-Cadherin (Shotgun) and Par-3 (Bazooka) levels at AJs. Maintaining E-Cadherin/Par-3 levels at the apical domain requires de novo synthesis rather than internalisation and recycling from and to the apical plasma membrane. However, apical targeting of E-Cadherin and Par-3 requires functional recycling endosomes, suggesting an intermediate role for this compartment in targeting de novo synthesized E-Cadherin to the plasma membrane. We demonstrate that apical enrichment of recycling endosomes is dependent on the MT motor Dynein and essential for the function of this vesicular compartment. In addition, we establish that E-Cadherin dynamics and MT requirement differ in remodelling tracheal cells versus planar epithelial cells. Altogether, our results uncover an MT-Dynein-dependent apical restriction of recycling endosomes that controls adhesion by sustaining Par-3 and E-Cadherin levels at AJs during morphogenesis.
Collapse
Affiliation(s)
- Pierre-Marie Le Droguen
- Institut Jacques Monod, CNRS and University Paris Diderot, 15 Rue H. Brion, Paris 75205, Cedex 13, France
| | - Sandra Claret
- Institut Jacques Monod, CNRS and University Paris Diderot, 15 Rue H. Brion, Paris 75205, Cedex 13, France
| | - Antoine Guichet
- Institut Jacques Monod, CNRS and University Paris Diderot, 15 Rue H. Brion, Paris 75205, Cedex 13, France
| | - Véronique Brodu
- Institut Jacques Monod, CNRS and University Paris Diderot, 15 Rue H. Brion, Paris 75205, Cedex 13, France
| |
Collapse
|
87
|
Satoh T, Ohba A, Liu Z, Inagaki T, Satoh AK. dPob/EMC is essential for biosynthesis of rhodopsin and other multi-pass membrane proteins in Drosophila photoreceptors. eLife 2015; 4. [PMID: 25715730 PMCID: PMC4341237 DOI: 10.7554/elife.06306] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 01/26/2015] [Indexed: 12/22/2022] Open
Abstract
In eukaryotes, most integral membrane proteins are synthesized, integrated into the membrane, and folded properly in the endoplasmic reticulum (ER). We screened the mutants affecting rhabdomeric expression of rhodopsin 1 (Rh1) in the Drosophila photoreceptors and found that dPob/EMC3, EMC1, and EMC8/9, Drosophila homologs of subunits of ER membrane protein complex (EMC), are essential for stabilization of immature Rh1 in an earlier step than that at which another Rh1-specific chaperone (NinaA) acts. dPob/EMC3 localizes to the ER and associates with EMC1 and calnexin. Moreover, EMC is required for the stable expression of other multi-pass transmembrane proteins such as minor rhodopsins Rh3 and Rh4, transient receptor potential, and Na+K+-ATPase, but not for a secreted protein or type I single-pass transmembrane proteins. Furthermore, we found that dPob/EMC3 deficiency induces rhabdomere degeneration in a light-independent manner. These results collectively indicate that EMC is a key factor in the biogenesis of multi-pass transmembrane proteins, including Rh1, and its loss causes retinal degeneration. DOI:http://dx.doi.org/10.7554/eLife.06306.001 The membranes that surround cells contain many proteins, and those that span the entire width of the membrane are known as transmembrane proteins. Rhodopsin is one such transmembrane protein that is found in the light-sensitive ‘photoreceptor’ cells of the eye, where it plays an essential role in vision. Transmembrane proteins are made inside the cell and are inserted into the membrane surrounding a compartment called the endoplasmic reticulum. Here, they mature and ‘fold’ into their correct three-dimensional shape with help from chaperone proteins. Once correctly folded, the transmembrane proteins can be transported to the cell membrane. Incorrect folding of proteins can have severe consequences; if rhodopsin is incorrectly folded the photoreceptor cells can die, leading to blindness in humans and other animals. Experiments carried out in zebrafish have shown that the chaperone protein Pob is required for the survival of photoreceptor cells. Pob is part of a group or ‘complex’ of chaperone proteins in the endoplasmic reticulum called the EMC complex. This suggests that the EMC complex may be involved in folding rhodopsin, but the details remain unclear. Here, Satoh et al. studied the role of the EMC complex in the folding of rhodopsin in fruit flies. This involved examining hundreds of flies that carried a variety of genetic mutations and that also had low levels of rhodopsin. The experiments show that dPob—the fly version of Pob—and two other proteins in the EMC complex are required for newly-made rhodopsin to be stabilized. If photoreceptor cells are missing proteins from the complex, the light-sensitive structures in the eye degenerate. Rhodopsin is known as a ‘multi-pass’ membrane protein because it crosses the membrane multiple times. Satoh et al. found that the EMC complex is also required for the folding of other multi-pass membrane proteins in photoreceptor cells. The next challenge will be to reveal how the EMC complex is able to specifically target this type of transmembrane protein. DOI:http://dx.doi.org/10.7554/eLife.06306.002
Collapse
Affiliation(s)
- Takunori Satoh
- Graduate School of Integrated Arts and Science, Hiroshima University, Higashi-Hiroshima, Japan
| | - Aya Ohba
- Graduate School of Integrated Arts and Science, Hiroshima University, Higashi-Hiroshima, Japan
| | - Ziguang Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, China
| | - Tsuyoshi Inagaki
- Graduate School of Integrated Arts and Science, Hiroshima University, Higashi-Hiroshima, Japan
| | - Akiko K Satoh
- Graduate School of Integrated Arts and Science, Hiroshima University, Higashi-Hiroshima, Japan
| |
Collapse
|
88
|
Chakrabarti P, Kolay S, Yadav S, Kumari K, Nair A, Trivedi D, Raghu P. A dPIP5K dependent pool of phosphatidylinositol 4,5 bisphosphate (PIP2) is required for G-protein coupled signal transduction in Drosophila photoreceptors. PLoS Genet 2015; 11:e1004948. [PMID: 25633995 PMCID: PMC4310717 DOI: 10.1371/journal.pgen.1004948] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/09/2014] [Indexed: 12/02/2022] Open
Abstract
Multiple PIP2 dependent molecular processes including receptor activated phospholipase C activity occur at the neuronal plasma membranes, yet levels of this lipid at the plasma membrane are remarkably stable. Although the existence of unique pools of PIP2 supporting these events has been proposed, the mechanism by which they are generated is unclear. In Drosophila photoreceptors, the hydrolysis of PIP2 by G-protein coupled phospholipase C activity is essential for sensory transduction of photons. We identify dPIP5K as an enzyme essential for PIP2 re-synthesis in photoreceptors. Loss of dPIP5K causes profound defects in the electrical response to light and light-induced PIP2 dynamics at the photoreceptor membrane. Overexpression of dPIP5K was able to accelerate the rate of PIP2 synthesis following light induced PIP2 depletion. Other PIP2 dependent processes such as endocytosis and cytoskeletal function were unaffected in photoreceptors lacking dPIP5K function. These results provide evidence for the existence of a unique dPIP5K dependent pool of PIP2 required for normal Drosophila phototransduction. Our results define the existence of multiple pools of PIP2 in photoreceptors generated by distinct lipid kinases and supporting specific molecular processes at neuronal membranes. PIP2 has been implicated in multiple functions at the plasma membrane. Some of these require its hydrolysis by receptor-activated phospholipase C, whereas others, such as membrane transport and cytoskeletal function, involve the interaction of the intact lipid with cellular proteins. The mechanistic basis underlying the segregation of these two classes of PIP2 dependent functions is unknown; it has been postulated that this might involve unique pools of PIP2 generated by distinct phosphoinsoitide kinases. We have studied this question in Drosophila photoreceptors, a model system where sensory transduction requires robust phospholipase C mediated PIP2 hydrolysis. We find that the activity of phosphatidylinositol-4-phosphate 5 kinase encoded by dPIP5K is required to support normal sensory transduction and PIP2 dynamics in photoreceptors. Remarkably, non-PLC dependent functions of PIP2, such as vesicular transport and the actin cytoskeleton, were unaffected in dPIP5K mutants. Thus, dPIP5K supports a pool of PIP2 that is readily available to PLC, but has no role in sustaining other non-PLC mediated PIP2 dependent processes. These findings support the existence of at least two non-overlapping pools of PIP2 at the plasma membrane, and provide a platform for future studies of PIP2 regulation at the plasma membrane.
Collapse
Affiliation(s)
| | - Sourav Kolay
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bangalore, India
- Manipal University, Madhav Nagar, Manipal, Karnataka, India
| | - Shweta Yadav
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bangalore, India
| | - Kamalesh Kumari
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bangalore, India
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Amit Nair
- Inositide Laboratory, Babraham Institute, Cambridge, United Kingdom
| | - Deepti Trivedi
- Inositide Laboratory, Babraham Institute, Cambridge, United Kingdom
| | - Padinjat Raghu
- Inositide Laboratory, Babraham Institute, Cambridge, United Kingdom
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bangalore, India
- * E-mail:
| |
Collapse
|
89
|
Farkaš R, Beňová-Liszeková D, Mentelová L, Mahmood S, Ďatková Z, Beňo M, Pečeňová L, Raška O, Šmigová J, Chase BA, Raška I, Mechler BM. Vacuole dynamics in the salivary glands ofDrosophila melanogasterduring prepupal development. Dev Growth Differ 2015; 57:74-96. [DOI: 10.1111/dgd.12193] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 10/21/2014] [Accepted: 11/28/2014] [Indexed: 01/29/2023]
Affiliation(s)
- Robert Farkaš
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
| | - Denisa Beňová-Liszeková
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
| | - Lucia Mentelová
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
- Department of Genetics; Comenius University; Mlynská dolina, B-1 84215 Bratislava Slovakia
| | - Silvia Mahmood
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
- Department of Medical Biochemistry; Jessenius Faculty of Medicine; Comenius University; Mala Hora 4 03601 Martin Slovakia
| | - Zuzana Ďatková
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
- Department of Genetics; Comenius University; Mlynská dolina, B-1 84215 Bratislava Slovakia
| | - Milan Beňo
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
| | - Ludmila Pečeňová
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
- Department of Genetics; Comenius University; Mlynská dolina, B-1 84215 Bratislava Slovakia
| | - Otakar Raška
- Institute of Cellular Biology and Pathology; 1st Faculty of Medicine; Charles University in Prague; Albertov 4 12800 Prague Czech Republic
| | - Jana Šmigová
- Institute of Cellular Biology and Pathology; 1st Faculty of Medicine; Charles University in Prague; Albertov 4 12800 Prague Czech Republic
| | - Bruce A. Chase
- Department of Biology; University of Nebraska at Omaha; 6001 Dodge Street Omaha NE 68182-0040 USA
| | - Ivan Raška
- Institute of Cellular Biology and Pathology; 1st Faculty of Medicine; Charles University in Prague; Albertov 4 12800 Prague Czech Republic
| | - Bernard M. Mechler
- Institute of Cellular Biology and Pathology; 1st Faculty of Medicine; Charles University in Prague; Albertov 4 12800 Prague Czech Republic
- German Cancer Research Centre; Neuenheimer Feld 581 D-69120 Heidelberg Germany
- VIT-University; Vellore Tamil Nadu India
| |
Collapse
|
90
|
Haltom AR, Lee TV, Harvey BM, Leonardi J, Chen YJ, Hong Y, Haltiwanger RS, Jafar-Nejad H. The protein O-glucosyltransferase Rumi modifies eyes shut to promote rhabdomere separation in Drosophila. PLoS Genet 2014; 10:e1004795. [PMID: 25412384 PMCID: PMC4238978 DOI: 10.1371/journal.pgen.1004795] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/02/2014] [Indexed: 12/31/2022] Open
Abstract
The protein O-glucosyltransferase Rumi/POGLUT1 regulates Drosophila Notch signaling by adding O-glucose residues to the Notch extracellular domain. Rumi has other predicted targets including Crumbs (Crb) and Eyes shut (Eys), both of which are involved in photoreceptor development. However, whether Rumi is required for the function of Crb and Eys remains unknown. Here we report that in the absence of Rumi or its enzymatic activity, several rhabdomeres in each ommatidium fail to separate from one another in a Notch-independent manner. Mass spectral analysis indicates the presence of O-glucose on Crb and Eys. However, mutating all O-glucosylation sites in a crb knock-in allele does not cause rhabdomere attachment, ruling out Crb as a biologically-relevant Rumi target in this process. In contrast, eys and rumi exhibit a dosage-sensitive genetic interaction. In addition, although in wild-type ommatidia most of the Eys protein is found in the inter-rhabdomeral space (IRS), in rumi mutants a significant fraction of Eys remains in the photoreceptor cells. The intracellular accumulation of Eys and the IRS defect worsen in rumi mutants raised at a higher temperature, and are accompanied by a ∼50% decrease in the total level of Eys. Moreover, removing one copy of an endoplasmic reticulum chaperone enhances the rhabdomere attachment in rumi mutant animals. Altogether, our data suggest that O-glucosylation of Eys by Rumi ensures rhabdomere separation by promoting proper Eys folding and stability in a critical time window during the mid-pupal stage. Human EYS, which is mutated in patients with autosomal recessive retinitis pigmentosa, also harbors multiple Rumi target sites. Therefore, the role of O-glucose in regulating Eys may be conserved.
Collapse
Affiliation(s)
- Amanda R. Haltom
- Program in Genes & Development, The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Tom V. Lee
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Beth M. Harvey
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Jessica Leonardi
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Yi-Jiun Chen
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Yang Hong
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Robert S. Haltiwanger
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Hamed Jafar-Nejad
- Program in Genes & Development, The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
91
|
Nucleotide bound to rab11a controls localization in rod cells but not interaction with rhodopsin. J Neurosci 2014; 34:14854-63. [PMID: 25378153 DOI: 10.1523/jneurosci.1943-14.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Precise vectorial transport of rhodopsin is essential for rod photoreceptor health and function. Mutations that truncate or extend the C terminus of rhodopsin disrupt this transport, and lead to retinal degeneration and blindness in human patients and in mouse models. Here we show that such mutations disrupt the binding of rhodopsin to the small GTPase rab11a. The rhodopsin-rab11a interaction is a direct binding interaction that does not depend on the nucleotide binding state of rab11a. Expression of EGFP-rab11a fusion proteins in Xenopus laevis photoreceptors revealed that the nucleotide binding status of rab11a affects its subcellular localization, with GTP-locked mutants concentrated in the inner segment and GDP-locked mutants concentrated in the outer segment. shRNA-mediated knockdown of rab11a in rods led to shortened outer segments and retinal degeneration. Together, our results show the critical importance of direct rhodopsin-rab11a interactions for the formation and maintenance of vertebrate photoreceptors.
Collapse
|
92
|
Tao YX, Conn PM. Chaperoning G protein-coupled receptors: from cell biology to therapeutics. Endocr Rev 2014; 35:602-47. [PMID: 24661201 PMCID: PMC4105357 DOI: 10.1210/er.2013-1121] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 03/14/2014] [Indexed: 12/13/2022]
Abstract
G protein-coupled receptors (GPCRs) are membrane proteins that traverse the plasma membrane seven times (hence, are also called 7TM receptors). The polytopic structure of GPCRs makes the folding of GPCRs difficult and complex. Indeed, many wild-type GPCRs are not folded optimally, and defects in folding are the most common cause of genetic diseases due to GPCR mutations. Both general and receptor-specific molecular chaperones aid the folding of GPCRs. Chemical chaperones have been shown to be able to correct the misfolding in mutant GPCRs, proving to be important tools for studying the structure-function relationship of GPCRs. However, their potential therapeutic value is very limited. Pharmacological chaperones (pharmacoperones) are potentially important novel therapeutics for treating genetic diseases caused by mutations in GPCR genes that resulted in misfolded mutant proteins. Pharmacoperones also increase cell surface expression of wild-type GPCRs; therefore, they could be used to treat diseases that do not harbor mutations in GPCRs. Recent studies have shown that indeed pharmacoperones work in both experimental animals and patients. High-throughput assays have been developed to identify new pharmacoperones that could be used as therapeutics for a number of endocrine and other genetic diseases.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology (Y.-X.T.), College of Veterinary Medicine, Auburn University, Auburn, Alabama 36849-5519; and Departments of Internal Medicine and Cell Biology (P.M.C.), Texas Tech University Health Science Center, Lubbock, Texas 79430-6252
| | | |
Collapse
|
93
|
Rab proteins: the key regulators of intracellular vesicle transport. Exp Cell Res 2014; 328:1-19. [PMID: 25088255 DOI: 10.1016/j.yexcr.2014.07.027] [Citation(s) in RCA: 203] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 07/06/2014] [Accepted: 07/23/2014] [Indexed: 01/01/2023]
Abstract
Vesicular/membrane trafficking essentially regulates the compartmentalization and abundance of proteins within the cells and contributes in many signalling pathways. This membrane transport in eukaryotic cells is a complex process regulated by a large and diverse array of proteins. A large group of monomeric small GTPases; the Rabs are essential components of this membrane trafficking route. Most of the Rabs are ubiquitously expressed proteins and have been implicated in vesicle formation, vesicle motility/delivery along cytoskeleton elements and docking/fusion at target membranes through the recruitment of effectors. Functional impairments of Rabs affecting transport pathways manifest different diseases. Rab functions are accompanied by cyclical activation and inactivation of GTP-bound and GDP-bound forms between the cytosol and membranes which is regulated by upstream regulators. Rab proteins are characterized by their distinct sub-cellular localization and regulate a wide variety of endocytic, transcytic and exocytic transport pathways. Mutations of Rabs affect cell growth, motility and other biological processes.
Collapse
|
94
|
Yashiro H, Loza AJ, Skeath JB, Longmore GD. Rho1 regulates adherens junction remodeling by promoting recycling endosome formation through activation of myosin II. Mol Biol Cell 2014; 25:2956-69. [PMID: 25079692 PMCID: PMC4230585 DOI: 10.1091/mbc.e14-04-0894] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Once adherens junctions (AJs) are formed between polarized epithelial cells they must be maintained because AJs are constantly remodeled in dynamic epithelia. AJ maintenance involves endocytosis and subsequent recycling of E-cadherin to a precise location along the basolateral membrane. In the Drosophila pupal eye epithelium, Rho1 GTPase regulates AJ remodeling through Drosophila E-cadherin (DE-cadherin) endocytosis by limiting Cdc42/Par6/aPKC complex activity. We demonstrate that Rho1 also influences AJ remodeling by regulating the formation of DE-cadherin-containing, Rab11-positive recycling endosomes in Drosophila postmitotic pupal eye epithelia. This effect of Rho1 is mediated through Rok-dependent, but not MLCK-dependent, stimulation of myosin II activity yet independent of its effects upon actin remodeling. Both Rho1 and pMLC localize on endosomal vesicles, suggesting that Rho1 might regulate the formation of recycling endosomes through localized myosin II activation. This work identifies spatially distinct functions for Rho1 in the regulation of DE-cadherin-containing vesicular trafficking during AJ remodeling in live epithelia.
Collapse
Affiliation(s)
- Hanako Yashiro
- ICCE Institute, Washington University School of Medicine, St. Louis, MO 63110 Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110 Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Andrew J Loza
- ICCE Institute, Washington University School of Medicine, St. Louis, MO 63110 Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
| | - James B Skeath
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Gregory D Longmore
- ICCE Institute, Washington University School of Medicine, St. Louis, MO 63110 Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110 Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110 BRIGHT Institute, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
95
|
Chung S, Andrew DJ. Cadherin 99C regulates apical expansion and cell rearrangement during epithelial tube elongation. Development 2014; 141:1950-60. [PMID: 24718992 DOI: 10.1242/dev.104166] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Apical and basolateral determinants specify and maintain membrane domains in epithelia. Here, we identify new roles for two apical surface proteins - Cadherin 99C (Cad99C) and Stranded at Second (SAS) - in conferring apical character in Drosophila tubular epithelia. Cad99C, the Drosophila ortholog of human Usher protocadherin PCDH15, is expressed in several embryonic tubular epithelial structures. Through loss-of-function and overexpression studies, we show that Cad99C is required to regulate cell rearrangement during salivary tube elongation. We further show that overexpression of either Cad99C or SAS causes a dramatic increase in apical membrane at the expense of other membrane domains, and that both proteins can do this independently of each other and independently of mislocalization of the apical determinant Crumbs (Crb). Overexpression of Cad99C or SAS results in similar, but distinct effects, suggesting both shared and unique roles for these proteins in conferring apical identity.
Collapse
Affiliation(s)
- Seyeon Chung
- Department of Cell Biology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205-2196, USA
| | | |
Collapse
|
96
|
Gurudev N, Yuan M, Knust E. chaoptin, prominin, eyes shut and crumbs form a genetic network controlling the apical compartment of Drosophila photoreceptor cells. Biol Open 2014; 3:332-41. [PMID: 24705015 PMCID: PMC4021355 DOI: 10.1242/bio.20147310] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The apical surface of epithelial cells is often highly specialised to fulfil cell type-specific functions. Many epithelial cells expand their apical surface by forming microvilli, actin-based, finger-like membrane protrusions. The apical surface of Drosophila photoreceptor cells (PRCs) forms tightly packed microvilli, which are organised into the photosensitive rhabdomeres. As previously shown, the GPI-anchored adhesion protein Chaoptin is required for the stability of the microvilli, whereas the transmembrane protein Crumbs is essential for proper rhabdomere morphogenesis. Here we show that chaoptin synergises with crumbs to ensure optimal rhabdomere width. In addition, reduction of crumbs ameliorates morphogenetic defects observed in PRCs mutant for prominin and eyes shut, known antagonists of chaoptin. These results suggest that these four genes provide a balance of adhesion and anti-adhesion to maintain microvilli development and maintenance. Similar to crumbs mutant PRCs, PRCs devoid of prominin or eyes shut undergo light-dependent retinal degeneration. Given the observation that human orthologues of crumbs, prominin and eyes shut result in progressive retinal degeneration and blindness, the Drosophila eye is ideally suited to unravel the genetic and cellular mechanisms that ensure morphogenesis of PRCs and their maintenance under light-mediated stress.
Collapse
Affiliation(s)
- Nagananda Gurudev
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, D-01307 Dresden, Germany
| | - Michaela Yuan
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, D-01307 Dresden, Germany
| | - Elisabeth Knust
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, D-01307 Dresden, Germany
| |
Collapse
|
97
|
Wang S, Tan KL, Agosto MA, Xiong B, Yamamoto S, Sandoval H, Jaiswal M, Bayat V, Zhang K, Charng WL, David G, Duraine L, Venkatachalam K, Wensel TG, Bellen HJ. The retromer complex is required for rhodopsin recycling and its loss leads to photoreceptor degeneration. PLoS Biol 2014; 12:e1001847. [PMID: 24781186 PMCID: PMC4004542 DOI: 10.1371/journal.pbio.1001847] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/21/2014] [Indexed: 12/22/2022] Open
Abstract
Rhodopsin mistrafficking can cause photoreceptor (PR) degeneration. Upon light exposure, activated rhodopsin 1 (Rh1) in Drosophila PRs is internalized via endocytosis and degraded in lysosomes. Whether internalized Rh1 can be recycled is unknown. Here, we show that the retromer complex is expressed in PRs where it is required for recycling endocytosed Rh1 upon light stimulation. In the absence of subunits of the retromer, Rh1 is processed in the endolysosomal pathway, leading to a dramatic increase in late endosomes, lysosomes, and light-dependent PR degeneration. Reducing Rh1 endocytosis or Rh1 levels in retromer mutants alleviates PR degeneration. In addition, increasing retromer abundance suppresses degenerative phenotypes of mutations that affect the endolysosomal system. Finally, expressing human Vps26 suppresses PR degeneration in Vps26 mutant PRs. We propose that the retromer plays a conserved role in recycling rhodopsins to maintain PR function and integrity.
Collapse
Affiliation(s)
- Shiuan Wang
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Kai Li Tan
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Melina A. Agosto
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Bo Xiong
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Shinya Yamamoto
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, United States of America
| | - Hector Sandoval
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Manish Jaiswal
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, Texas, United States of America
| | - Vafa Bayat
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ke Zhang
- Program in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Wu-Lin Charng
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Gabriela David
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lita Duraine
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, Texas, United States of America
| | - Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, University of Texas School of Medicine, Houston, Texas, United States of America
| | - Theodore G. Wensel
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hugo J. Bellen
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, United States of America
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
98
|
Jones TA, Nikolova LS, Schjelderup A, Metzstein MM. Exocyst-mediated membrane trafficking is required for branch outgrowth in Drosophila tracheal terminal cells. Dev Biol 2014; 390:41-50. [PMID: 24607370 DOI: 10.1016/j.ydbio.2014.02.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 02/19/2014] [Accepted: 02/20/2014] [Indexed: 11/16/2022]
Abstract
Branching morphogenesis, the process by which cells or tissues generate tree-like networks that function to increase surface area or in contacting multiple targets, is a common developmental motif in multicellular organisms. We use Drosophila tracheal terminal cells, a component of the insect respiratory system, to investigate branching morphogenesis that occurs at the single cell level. Here, we show that the exocyst, a conserved protein complex that facilitates docking and tethering of vesicles at the plasma membrane, is required for terminal cell branch outgrowth. We find that exocyst-deficient terminal cells have highly truncated branches and show an accumulation of vesicles within their cytoplasm and are also defective in subcellular lumen formation. We also show that vesicle trafficking pathways mediated by the Rab GTPases Rab10 and Rab11 are redundantly required for branch outgrowth. In terminal cells, the PAR-polarity complex is required for branching, and we find that the PAR complex is required for proper membrane localization of the exocyst, thus identifying a molecular link between the branching and outgrowth programs. Together, our results suggest a model where exocyst mediated vesicle trafficking facilitates branch outgrowth, while de novo branching requires cooperation between the PAR and exocyst complexes.
Collapse
Affiliation(s)
- Tiffani A Jones
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Linda S Nikolova
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Ani Schjelderup
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Mark M Metzstein
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
99
|
Four-dimensional live imaging of apical biosynthetic trafficking reveals a post-Golgi sorting role of apical endosomal intermediates. Proc Natl Acad Sci U S A 2014; 111:4127-32. [PMID: 24591614 DOI: 10.1073/pnas.1304168111] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Emerging data suggest that in polarized epithelial cells newly synthesized apical and basolateral plasma membrane proteins traffic through different endosomal compartments en route to the respective cell surface. However, direct evidence for trans-endosomal pathways of plasma membrane proteins is still missing and the mechanisms involved are poorly understood. Here, we imaged the entire biosynthetic route of rhodopsin-GFP, an apical marker in epithelial cells, synchronized through recombinant conditional aggregation domains, in live Madin-Darby canine kidney cells using spinning disk confocal microscopy. Our experiments directly demonstrate that rhodopsin-GFP traffics through apical recycling endosomes (AREs) that bear the small GTPase Rab11a before arriving at the apical membrane. Expression of dominant-negative Rab11a drastically reduced apical delivery of rhodopsin-GFP and caused its missorting to the basolateral membrane. Surprisingly, functional inhibition of dynamin-2 trapped rhodopsin-GFP at AREs and caused aberrant accumulation of coated vesicles on AREs, suggesting a previously unrecognized role for dynamin-2 in the scission of apical carrier vesicles from AREs. A second set of experiments, using a unique method to carry out total internal reflection fluorescence microscopy (TIRFM) from the apical side, allowed us to visualize the fusion of rhodopsin-GFP carrier vesicles, which occurred randomly all over the apical plasma membrane. Furthermore, two-color TIRFM showed that Rab11a-mCherry was present in rhodopsin-GFP carrier vesicles and was rapidly released upon fusion onset. Our results provide direct evidence for a role of AREs as a post-Golgi sorting hub in the biosynthetic route of polarized epithelia, with Rab11a regulating cargo sorting at AREs and carrier vesicle docking at the apical membrane.
Collapse
|
100
|
Alvers AL, Ryan S, Scherz PJ, Huisken J, Bagnat M. Single continuous lumen formation in the zebrafish gut is mediated by smoothened-dependent tissue remodeling. Development 2014; 141:1110-9. [PMID: 24504339 DOI: 10.1242/dev.100313] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The formation of a single lumen during tubulogenesis is crucial for the development and function of many organs. Although 3D cell culture models have identified molecular mechanisms controlling lumen formation in vitro, their function during vertebrate organogenesis is poorly understood. Using light sheet microscopy and genetic approaches we have investigated single lumen formation in the zebrafish gut. Here we show that during gut development multiple lumens open and enlarge to generate a distinct intermediate, which consists of two adjacent unfused lumens separated by basolateral contacts. We observed that these lumens arise independently from each other along the length of the gut and do not share a continuous apical surface. Resolution of this intermediate into a single, continuous lumen requires the remodeling of contacts between adjacent lumens and subsequent lumen fusion. We show that lumen resolution, but not lumen opening, is impaired in smoothened (smo) mutants, indicating that fluid-driven lumen enlargement and resolution are two distinct processes. Furthermore, we show that smo mutants exhibit perturbations in the Rab11 trafficking pathway and demonstrate that Rab11-mediated trafficking is necessary for single lumen formation. Thus, lumen resolution is a distinct genetically controlled process crucial for single, continuous lumen formation in the zebrafish gut.
Collapse
Affiliation(s)
- Ashley L Alvers
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|